Elsevier

Life Sciences

Volume 241, 15 January 2020, 117146
Life Sciences

HMGB1 upregulates NF-kB by inhibiting IKB-α and associates with diabetic retinopathy

https://doi.org/10.1016/j.lfs.2019.117146Get rights and content

Abstract

Aims

Diabetic retinopathy (DR) is the main cause of blindness in adults and investigating new therapeutic targets for DR is necessary. This study aimed to investigate the effect of high-mobility group box 1 (HMGB1) protein and its mechanism in diabetic retinopathy (DR) were investigated.

Main methods

Human retinal endothelial cells (HREC) were uesd for chip-seq. Sprague Dawley (SD) rats were randomly divided into control group, HMGB1 group, diabetes mellitus (DM) combined with HMGB1 siRNA group, and DM group. Next, eyeballs were removed and retinas were detached for western blot. The DM model of cell was built by increasing the glucose concentration in cell culture medium. The regulation of HMGB1 was achieved by short hairpin (sh)-HMGB1 transfection, then, the transfected cells were harvested for luciferase assay, western blot and qRT-PCR analyses as well as proliferation and apoptosis detection.

Key findings

Chip-seq and luciferase assay showed the possible transcription factor functions of HMGB1 and IKB-α was one of the HMGB1 binding sites. In vivo and in vitro results indicated high expression of HMGB1 and NF-kB and low expression of IKB-α in DR and the expression of IKB-α and NF-kB was regulated by HMGB1. Moreover, cell assays showed that HMGB1 inhibited cell proliferation and promoted apoptosis.

Significance

The results from the present study showed that HMGB1 may be involved in the pathogenesis of DR as a transcription factor through NF-kB pathway. Therefore, blockade of HMGB1 may be a new method for the treatment of DR.

Introduction

Diabetic retinopathy (DR), a major microvascular complication of diabetes mellitus (DM), is the main cause of blindness in adults [[1], [2], [3]]. Because the extremely complicated pathogenesis of DR has not been fully elucidated, investigating and finding new therapeutic targets for DR is necessary. The main pathological changes of DR include retinal cell apoptosis and angiogenesis [4]. Previous studies have shown that progression of DR is accompanied by oxidative stress [5], mitochondrial dysfunction [6], and inflammation [7] as well as other clinical manifestations.

High-mobility group box 1 (HMGB1) protein is a highly conserved non-histone DNA-binding protein that plays an important role in many biological behaviors. As a late inflammatory factor, HMGB1 can be released into extracellular fluid actively or passively after stress [8,9]. In the cytoplasm, HMGB1 is involved in the imbalance of autophagy and apoptosis [10]. When released, HMGB1 can regulate angiogenesis [11], cell metastasis, and release of inflammatory cytokines [[12], [13], [14]].

Recent studies [15,16] have shown that HMGB1 is associated with diabetic peripheral neuropathy and involved in the occurrence and progression of DR; however, the underlying mechanism remains unclear. Therefore, in the present study, the specific effects of HMGB1 and its mechanism in DR were investigated.

Section snippets

Animals and groups

Forty specific-pathogen-free (SPF) male Sprague Dawley (SD) rats weighing 240–280 g were selected and randomly divided into the normal or DM group. All animal experiments were approved by the Ethics Committee of the Affiliated Hospital of China Medical University (ethics number: 2016PS229K). Rats in the DM group were injected intraperitoneally with 1% streptozototocin (STZ, Sigma, USA) at the dose of 60 mg/kg, and the rats in the normal group were injected intraperitoneally with an equal amount

Animal model

Weight and blood glucose were not significantly different among the control, HMGB1, DM combined with HMGB1 siRNA, and DM groups (p > 0.05). After 72 h of injection with STZ, the blood glucose in rats in both the DM combined with HMGB1 siRNA and DM groups increased to >16.7 mmol/L, indicating the establishment of the DM model was 100% successful. By comparison, the weight of rats in the DM combined with HMGB1 siRNA and DM groups was lower than in the control and HMGB1 groups (all p < 0.05);

Proliferation and apoptosis

After the overexpression of HMGB1, CCK-8 was used to investigate the impact of HMGB1 on proliferation of the HREC line. Cell proliferation in the sh-HMGB1 transfection group was lower than in the sh-NC transfection group (Fig. 4B). Flow cytometry results showed the apoptosis rate in the sh-HMGB1 transfection group was higher than in the sh-NC transfection group (Fig. 4A).

Discussion

HMGB1, a multifunctional protein, is associated with a variety of physiological and pathological processes, including cell proliferation and differentiation, inflammation, immunity, cancer, metabolism, and oxidative stress [[17], [18], [19], [20], [21]]. However, information regarding the function of HMGB1 in DR is relatively limited. In the present study, the HMGB1 expression in the retinas of diabetic rats at 17 weeks of age was higher than in the control group. After HREC were treated for

Conclusions

In conclusion, the results from this study showed that HMGB1 may affected the NF-kB pathway through IKB-α, thus affecting the pathogenesis of DR, and the inhibition of HMGB1 may provide a novel concept for the treatment of DR.

The following are the supplementary data related to this article.

. HMGB1 binding sites

Funding

This work was partly supported by the National Natural Science Foundation of China (81570866).

Ethics approval and consent to participate

Animal experimental procedures were in accordance with institutional guidelines and approved by the Ethics Committee of the Affiliated Hospital of China Medical University (ethics number: 2016PS229K).

Declaration of competing interest

The authors declare that there are no conflicts of interest.

References (37)

  • P.F. Torres et al.

    The role of cytokines in corneal immunopathology

    Ocul. Immunol. Inflamm.

    (2001)
  • R.C. Chen

    The role of HMGB1-RAGE axis in migration and invasion of hepatocellular carcinoma cell lines

    Mol. Cell. Biochem.

    (2014)
  • W.K. Kim

    Identification of specifically activated angiogenic molecules in HMGB-1-induced angiogenesis

    BMB Rep.

    (2017)
  • X. Zhu

    Cytosolic HMGB1 controls the cellular autophagy/apoptosis checkpoint during inflammation

    J. Clin. Invest.

    (2015)
  • G.P. Sims et al.

    HMGB1 and RAGE in inflammation and cancer

    Annu. Rev. Immunol.

    (2010)
  • U. Andersson et al.

    HMGB1 is a therapeutic target for sterile inflammation and infection

    Annu. Rev. Immunol.

    (2011)
  • J.O. Thomas et al.

    H1 and HMGB1: modulators of chromatin structure

    Biochem. Soc. Trans.

    (2012)
  • H. Zhao et al.

    HMGB-1 as a potential target for the treatment of diabetic retinopathy

    Med. Sci. Monit.

    (2015)
  • Cited by (74)

    View all citing articles on Scopus
    View full text